Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 9: 453, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29563915

RESUMO

Complement receptor type 1 (CR1) is a multi modular membrane receptor composed of 30 homologous complement control protein modules (CCP) organized in four different functional regions called long homologous repeats (LHR A, B, C, and D). CR1 is a receptor for complement-opsonins C3b and C4b and specifically interacts through pairs of CCP modules located in LHR A, B, and C. Defense collagens such as mannose-binding lectin (MBL), ficolin-2, and C1q also act as opsonins and are involved in immune clearance through binding to the LHR-D region of CR1. Our previous results using deletion variants of CR1 mapped the interaction site for MBL and ficolin-2 on CCP24-25. The present work aimed at deciphering the interaction of C1q with CR1 using new CR1 variants concentrated around CCP24-25. CR1 bimodular fragment CCP24-25 and CR1 CCP22-30 deleted from CCP24-25 produced in eukaryotic cells enabled to highlight that the interaction site for both MBL and C1q is located on the same pair of modules CCP24-25. C1q binding to CR1 shares with MBL a main common interaction site on the collagen stalks but also subsidiary sites most probably located on C1q globular heads, contrarily to MBL.


Assuntos
Complemento C1q/química , Lectina de Ligação a Manose/química , Peptídeos/química , Receptores de Complemento 3b/química , Complemento C1q/genética , Complemento C1q/imunologia , Humanos , Lectinas/química , Lectinas/genética , Lectinas/imunologia , Lectina de Ligação a Manose/genética , Lectina de Ligação a Manose/imunologia , Peptídeos/genética , Peptídeos/imunologia , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/imunologia , Ficolinas
2.
Mol Immunol ; 67(2 Pt B): 584-95, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26260209

RESUMO

The regulators of complement activation gene cluster encodes a group of proteins that have evolved to control the amplification of complement at the critical step of C3 activation. Complement receptor 1 (CR1) is the most versatile of these inhibitors with both receptor and regulatory functions. While expressed on most peripheral blood cells, the only epithelial site of expression in the kidney is by the podocyte. Its expression by this cell population has aroused considerable speculation as to its biologic function in view of many complement-mediated renal diseases. The goal of this investigation was to assess the role of CR1 on epithelial cells. To this end, we utilized a Chinese hamster ovary cell model system. Among our findings, CR1 reduced C3b deposition by ∼ 80% during classical pathway activation; however, it was an even more potent regulator (>95% reduction in C3b deposition) of the alternative pathway. This inhibition was primarily mediated by decay accelerating activity. The deposited C4b and C3b were progressively cleaved with a t½ of ∼ 30 min to C4d and C3d, respectively, by CR1-dependent cofactor activity. CR1 functioned intrinsically (i.e, worked only on the cell on which it was expressed). Moreover, CR1 efficiently and stably bound but didn't internalize C4b/C3b opsonized immune complexes. Our studies underscore the potential importance of CR1 on an epithelial cell population as both an intrinsic complement regulator and an immune adherence receptor. These results provide a framework for understanding how loss of CR1 expression on podocytes may contribute to complement-mediated damage in the kidney.


Assuntos
Proteínas do Sistema Complemento/imunologia , Células Epiteliais/metabolismo , Rim/metabolismo , Rim/patologia , Modelos Imunológicos , Receptores de Complemento 3b/metabolismo , Animais , Células CHO , Complemento C4b , Via Alternativa do Complemento/imunologia , Cricetinae , Cricetulus , Eritrócitos/metabolismo , Humanos , Cinética , Proteínas Opsonizantes/metabolismo , Receptores de Complemento 3b/química , Transfecção
3.
J Biol Chem ; 284(51): 35605-11, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19833734

RESUMO

CRIg is a recently discovered complement C3 receptor expressed on a subpopulation of tissue-resident macrophages. The extracellular IgV domain of CRIg (CRIg-ECD) holds considerable promise as a potential therapeutic because it selectively inhibits the alternative pathway of complement by binding to C3b and inhibiting proteolytic activation of C3 and C5. However, CRIg binds weakly to the convertase subunit C3b (K(D) = 1.1 microm), and thus a relatively high concentration of protein is required to reach nearly complete complement inhibition. To improve therapeutic efficacy while minimizing risk of immunogenicity, we devised a phage display strategy to evolve a high affinity CRIg-ECD variant with a minimal number of mutations. Using the crystal structure of CRIg in complex with C3b as a guide for library design, we isolated a CRIg-ECD double mutant (Q64R/M86Y, CRIg-v27) that showed increased binding affinity and improved complement inhibitory activity relative to CRIg-ECD. In a mouse model of arthritis, treatment with a Fc fusion of CRIg-v27 resulted in a significant reduction in clinical scores compared with treatment with an Fc fusion of CRIg-ECD. This study clearly illustrates how phage display technology and structural information can be combined to generate proteins with nearly natural sequences that act as potent complement inhibitors with greatly improved therapeutic efficacy.


Assuntos
Artrite/tratamento farmacológico , Receptores de Complemento 3b/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Substituição de Aminoácidos , Animais , Artrite/metabolismo , Complemento C3b/genética , Complemento C3b/metabolismo , Complemento C5/genética , Complemento C5/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína/fisiologia , Coelhos , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Relação Estrutura-Atividade
4.
Mol Immunol ; 45(16): 4041-7, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18752851

RESUMO

Complement receptor of the immunoglobulin superfamily (CRIg), also referred to as Z39Ig and V-set and Ig domain-containing 4 (VSIG4), has recently been implicated in the clearance of systemic pathogens and autologous cells. CRIg is exclusively expressed on tissue resident macrophages and binds to multimers of C3b and iC3b that are covalently attached to particle surfaces. Next to functioning as an important clearance receptor, CRIg's extracellular domain inhibits complement activation through the alternative, but not the classical, pathway, providing a novel tool to selectively block this pathway in vivo. Here, we review a role for CRIg in immune clearance, T-cell responses and complement regulation, and discuss the implications for disease manifestation.


Assuntos
Inflamação/imunologia , Macrófagos/imunologia , Receptores de Complemento 3b/imunologia , Receptores de Complemento/imunologia , Animais , Humanos , Macrófagos/metabolismo , Modelos Imunológicos , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/metabolismo
5.
Am J Hematol ; 82(11): 970-5, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17654505

RESUMO

Immune complexes (IC) are constantly formed at low levels in normal individuals. In humans, the red blood cell (RBC) complement receptor 1 (CR1) plays the dominant role in the IC binding and clearance. Over the last several years, we have investigated the potential utility of immunocamouflaged (methoxypoly(ethylene glycol) [mPEG] grafted) RBC to attenuate the risk of alloimmunization. Because the grafted polymer nonspecifically camouflages membrane proteins, its effects on CR1 detection and IC binding were assessed. The dose dependent (0-2.5 mM) effects of activated mPEG (CmPEG, 5 kDa; and BTCmPEG, 5 and 20 kDa) on CR1 detection and the binding of artificially generated IC [C3b coated alkaline phosphatase and antialkaline phosphatase complexes] to control and pegylated RBC was investigated by flow cytometry. In contrast to selected non-ABO blood group antigens, grafted mPEG did not effectively camouflage CR1. Surprisingly, however, even very low grafting concentrations of mPEG (>or=0.3 mM) resulted in a >or=95% loss in IC binding. Further reductions in grafting concentration (0.15 and 0.03 mM mPEG) still yielded decreased IC binding of approximately 60 and 40%, respectively. Importantly, unactivated mPEG had minimal effects on IC binding. These data demonstrate that even small amounts of grafted mPEG interfere with the multivalent CR1-IC interaction necessary for high affinity IC binding, hence large volume transfusions of mPEG-RBC may be contraindicated in patients with pre-existing IC disease. Whether this concern is of clinical significance in healthy humans is less clear due to dilutional effects and the presence of secondary clearance pathways.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Incompatibilidade de Grupos Sanguíneos/prevenção & controle , Transfusão de Eritrócitos/métodos , Eritrócitos/imunologia , Polietilenoglicóis/química , Sistema ABO de Grupos Sanguíneos/imunologia , Complexo Antígeno-Anticorpo/química , Incompatibilidade de Grupos Sanguíneos/imunologia , Eritrócitos/química , Humanos , Técnicas In Vitro , Receptores de Complemento 3b/química , Receptores de Complemento 3b/imunologia , Sistema do Grupo Sanguíneo Rh-Hr/imunologia
6.
Biochem Biophys Res Commun ; 353(2): 363-368, 2007 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-17178110

RESUMO

Complement sensitization of red blood cells (RBCs) can cause life-threatening hemolytic anemias. We have previously shown that complement receptor 1 (CR1) derivatives specifically the N-terminal region with decay accelerating activity (DAA) for inactivation of a key enzyme in the complement cascade can reduce complement-mediated RBC destruction in vitro and in an in vivo mouse model of hemolytic transfusion reaction. In the present study, we have modeled the N-terminal CR1 molecule based on the X-ray crystal structure of decay accelerating factor and the NMR structure of a homologous CR1 domain. Based on the homology model, we identified a 34-mer peptide encompassing the putative DAA which in vitro reduced hemolysis, C3a release and surface C3 deposition. More importantly, this peptide at 0.6 mM was effective in prolonging survival of transfused incompatible RBCs in vivo. Our results indicate that CR1-based structure-function studies may provide insights for developing structure-derived transfusion therapeutics in the future.


Assuntos
Hemólise/imunologia , Modelos Químicos , Modelos Moleculares , Peptídeos/química , Peptídeos/imunologia , Receptores de Complemento 3b/química , Receptores de Complemento 3b/imunologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Simulação por Computador , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Receptores de Complemento/química , Receptores de Complemento/imunologia , Homologia de Sequência de Aminoácidos
7.
Am J Pathol ; 163(4): 1457-65, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14507653

RESUMO

Complement activation during ischemia and reperfusion contributes to the development of tissue injury with severe negative impact on outcomes in transplantation. To counter the effect of complement, we present a strategy to deliver a novel complement regulator stabilized on cell surfaces within donor organs. The membrane-bound complement regulator is able to inhibit complement activation when the donor organ is revascularized and exposed to host-circulating complement. Application of this construct to donor kidneys protected transplanted tissues from ischemia/reperfusion injury and reduced the deposition of activated complement and histological signs of damage under conditions in which a nontargeted control construct was ineffective. Treatment of donor organs in this way improved graft performance in the short and long term. An analysis of the immune response in allograft recipients showed that reducing graft damage at the time of transplantation through complement regulation also modulated the alloresponse. Additionally, the results of perfusion studies with human kidneys demonstrated the feasibility of targeting endothelial and epithelial surfaces with this construct, to allow investigation in clinical transplantation.


Assuntos
Transplante de Rim , Rim/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Complemento 3b/química , Circulação Renal , Traumatismo por Reperfusão/prevenção & controle , Doença Aguda , Animais , Doença Crônica , Endotélio/metabolismo , Endotélio/patologia , Células Epiteliais/metabolismo , Rejeição de Enxerto/fisiopatologia , Rejeição de Enxerto/prevenção & controle , Humanos , Técnicas In Vitro , Rim/metabolismo , Rim/fisiopatologia , Nefropatias/prevenção & controle , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Receptores de Superfície Celular/metabolismo , Doadores de Tecidos
8.
J Gen Virol ; 84(Pt 5): 1091-1100, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12692273

RESUMO

Simian herpes B virus (SHBV) is the herpes simplex virus (HSV) homologue for the species MACACA: Unlike in its natural host, and unlike other animal herpesviruses, SHBV causes high mortality in accidentally infected humans. SHBV-infected cells, like those infected with HSV-1 and equine herpesvirus types 1 and 4, express complement C3 receptor activity. To study immunoregulatory functions involved in susceptibility/resistance against interspecies transmission, the SHBV glycoprotein C (gC(SHBV)) gene (encoding 467 aa) was isolated. Sequence analysis revealed amino acid identity with gC proteins from HSV-2 (46.9 %), HSV-1 (44.5 %) and pseudorabies virus (21.2 %). Highly conserved cysteine residues were also noted. Similar to gC(HSV-2), gC(SHBV) is less glycosylated than gC(HSV-1), resulting in a molecular mass of 65 kDa if expressed in replication-deficient vaccinia virus Ankara. Stable transfectants expressing full-length gC(SHBV) on the cell surface induced C3 receptor activity and were substantially protected from complement-mediated lysis; no protection was observed with control constructs. This suggests that expression of the gC homologues on infected cell surfaces might also contribute to the survival of infected cells in addition to decreased virion inactivation. Interestingly, soluble gC(SHBV) isolated from protein-free culture supernatants did not interfere with the binding of the alternative complement pathway activator properdin to C3b, which is similar to our findings with gC(HSV-2) and could be attributed to major differences in the amino-terminal portion of the protein with extended deletions in both gC(SHBV) and gC(HSV-2). Binding of recombinant gC(SHBV) to polysulphates was observed. This, together with the heparin-sensitivity of the gC(SHBV)-C3 interaction on the infected cell surface, suggests a role in adherence to heparan sulphate, similar to the gC proteins of other herpesviruses.


Assuntos
Clonagem Molecular , Complemento C3b/imunologia , Herpesvirus Cercopitecino 1/patogenicidade , Receptores de Complemento 3b/metabolismo , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Via Alternativa do Complemento/imunologia , Citotoxicidade Imunológica , Humanos , Dados de Sequência Molecular , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Análise de Sequência de DNA , Transfecção , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
9.
Ann N Y Acad Sci ; 938: 196-206; discussion 206-7, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11458509

RESUMO

Current data suggest that interplay between two classes of molecules contributes to the regulation of hematopoiesis: hematopoietic growth factors, which regulate the survival, proliferation, and development of primitive hematopoietic cells and cell adhesion molecules (CAMs), which are responsible for the localization of hematopoiesis to the bone marrow (BM) and for mediating physical association between developing hematopoietic cells and marrow stromal tissue. A range of cell surface molecules representing several CAM superfamilies including integrins, selectins, the immunoglobulin gene superfamily and an emerging family of mucin-like molecules (the sialomucins) are involved in supporting cell-cell and cell-extracellular matrix (ECM) interactions between primitive hematopoietic cells and the stromal cell-mediated hematopoietic microenvironment (HM) of the bone marrow. There is abundant evidence in non-hematopoietic tissues that CAMs are signalling molecules which participate in a range of signal transduction events important not only for regulating cell adhesion and motility, but also for cell growth and survival. Although the signalling functions of CAMs have not been studied extensively in primitive hematopoietic progenitors (HPCs), extrapolation from burgeoning data in other systems is consistent with the hypothesis that hematopoiesis within the BM is regulated by interaction between signals generated locally by CAMs and those elicited by cytokines. Evidence in support of this notion was initially provided by studies on normal HPCs demonstrating cross-talk between members of the integrin superfamily and cytokine receptors. In this article we review recent reports that mucin-like molecules are also signalling molecules on primitive hematopoietic cells and that the signals they deliver potently inhibit hematopoiesis.


Assuntos
Antígenos CD , Moléculas de Adesão Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Mucinas/fisiologia , Moléculas de Adesão de Célula Nervosa , Animais , Antígeno CD146 , Moléculas de Adesão Celular/química , Divisão Celular , Sobrevivência Celular , Selectina E/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucossialina , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Mucinas/química , Mucinas/genética , Selectina-P/fisiologia , Estrutura Terciária de Proteína , Receptores de Superfície Celular/química , Receptores de Superfície Celular/fisiologia , Receptores de Complemento 3b/química , Receptores de Complemento 3b/fisiologia , Sialoglicoproteínas/química , Sialoglicoproteínas/fisiologia , Sialomucinas , Células Estromais/citologia
10.
Curr Protoc Immunol ; Chapter 13: 13.5.1-13.5.18, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18432718

RESUMO

The complement system is the primary effector of humoral immunity. Because of its enormous destructive capacity, mechanisms for confining the activity of the system to the desired target and elaborate safeguards for protecting self against complement-mediated injury have evolved. Human cells, particularly those found at sites of inflammation (e.g., hematopoietic and endothelial cells), express highly specialized membrane constituents that act independently or in concert with plasma regulatory proteins to inhibit the functional activity of complement. Decay-accelerating factor (DAF), or CD55, directly inhibits the formation and stability of the amplification C3 and C5 convertases of both the classical and the alternative pathways. Failure of a cell to regulate the amplification C3 and C5 convertases allows the generation of the potentially cytolytic membrane attack complex (MAC), or C5b-9 (consisting of the complement components C5b, C6, C7, C8, and C9). The primary cellular regulator of the MAC is the membrane inhibitor of reactive lysis (MIRL), or CD59, which restricts complement-mediated lysis by blocking assembly of the MAC (primarily at the stage of C9 binding and polymerization). This unit provides a basic protocol for isolating CD55 and CD59, along with two support protocols describing separate functional assays for CD59 and CD55.


Assuntos
Antígenos CD55/análise , Antígenos CD55/isolamento & purificação , Antígenos CD59/análise , Antígenos CD59/isolamento & purificação , Antígenos CD55/imunologia , Antígenos CD59/imunologia , Cromatografia Líquida/métodos , Humanos , Proteína Cofatora de Membrana/química , Proteína Cofatora de Membrana/imunologia , Receptores de Complemento 3b/química , Receptores de Complemento 3b/imunologia , Sensibilidade e Especificidade
11.
Mol Immunol ; 34(4): 297-304, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9244342

RESUMO

Complement receptor type one (CR1) in primates has several remarkable structural features including a size polymorphism (Mr 190000, 220000, 250000 and 280000) in man, multiple size variants (Mr 55000-220000) among non-human primates, and a partial amino-terminal duplication (CR1-like gene) that appears to encode the short (55000-70000) forms expressed on primate erythrocytes. In general, these short CR1 forms, some of which are GPI anchored, are expressed on erythrocytes and the 220000 molecular weight CR1 form is expressed on PBMC, except in man, where only the 220000 molecular weight form has been detected. In addition, the Mr 220000 human CR1 sequence carries several long internal repeats of up to 99% homology. It has been suggested that the highest homology is maintained by gene conversion and/or unequal crossover. To address further the evolutionary and biologic implications of these multiple forms, a 6 kb cDNA encoding baboon CR1(220) was identified by RTPCR using human CR1 primers. Its sequence contains the expected 30 complement control protein repeats (CCP) and demonstrates an overall homology to human CR1 of 95.4% at the nucleotide level and 93.2% at the amino acid level. As in human CR1, the first 28 CCP maintain the characteristic "seven CCP-long homologous repeats (LHR)" organization. Analysis of baboon CR1(220) indicates that horizontal or concerted evolution has maintained a high degree (> 98%) of identity between corresponding CCP within the LHRs from CCP 4 to CCP 19, while this homology region extends from CCP 3 to CCP 18 in man. In contrast, substitutions occurring in other CCP are not propagated to the corresponding sites of other LHR. Sequence differences in CCP 1, 2 and 3 are likely to be related to the acquisition of enhanced C3b binding capability by this amino-terminal region of the protein. Thus, the sequence data strongly support the hypotheses that gene conversion and or unequal crossover events have driven the evolution of the protein in regions of high homology while selective forces, probably ligand binding requirements, have maintained the regions of divergence.


Assuntos
Evolução Molecular , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Sequência de Aminoácidos , Animais , Linhagem Celular Transformada , Transformação Celular Viral , DNA Complementar/química , DNA Complementar/isolamento & purificação , Herpesvirus Humano 4 , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ligantes , Dados de Sequência Molecular , Papio , Ligação Proteica/imunologia , Receptores de Complemento 3b/biossíntese , Sequências Repetitivas de Ácido Nucleico , Homologia de Sequência de Aminoácidos
12.
Autoimmunity ; 23(1): 1-8, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8871757

RESUMO

Experimental autoimmune thyroiditis (EAT), induced by immunisation of rats with thyroid extract and complete Freund's adjuvant, has been used as a model to study the effects of complement inhibition mediated by soluble complement receptor 1 (sCR1) administration during the initial phase of the disease. There was no effect of sCR1 on the severity of thyroiditis at day 28 after immunisation or on the levels of thyroid antibodies, whether sCR1 was given during the first or second week after immunisation. Human IgG containing high levels of thyroid peroxidase antibodies given to rats at the time of immunisation caused significant worsening of thyroiditis severity (P < 0.01 compared to animals receiving normal IgG) but sCR1 again had no effect in this variant of the EAT model. The results indicate that complement does not play a major role in the initial phase of tissue injury in EAT and complement inhibition does not impair the generation of an autoimmune response against the thyroid, although it remains possible that complement activation is important during the chronic phase of disease maintenance in human autoimmune thyroid disease.


Assuntos
Autoanticorpos/imunologia , Receptores de Complemento 3b/imunologia , Glândula Tireoide/imunologia , Tireoidite Autoimune/imunologia , Animais , Ativação do Complemento , Feminino , Humanos , Ratos , Ratos Endogâmicos , Receptores de Complemento 3b/química , Solubilidade
13.
Protein Expr Purif ; 6(6): 727-36, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8746624

RESUMO

We have developed a simple expression, isolation, and folding protocol for an SCR oligomer comprising the first three SCRs of complement receptor Type 1 (C3b/C4b receptor, CD35). A T7 RNA polymerase expression system in Escherichia coli was used to express the oligomer as inclusion bodies. The oligomer was recovered from solubilized inclusion bodies using batch adsorption on SP-Sepharose. The oligomer was folded by one-step dilution in 20 mM ethanolamine/1 mM EDTA supplemented with 1 mM GSH/0.5 mM GSSG. The folded material was processed to a concentrated (> 20 mg/ml), usable product of greater than 98% purity using a combination of ultrafiltration, ammonium sulfate treatment, hydrophobic interaction, and size-exclusion chromatography. The yield of folded material varied between 6 and 15 mg/liter culture. The oxidation states of the 12 cysteine residues in SCR(1-3) were identified by HPLC of peptide fragments from a tryptic digest using dual UV/fluorescence detection, collection of selected peaks, and N-terminal sequencing. This methodology confirmed the expected location of disulfide bridges. Equilibrium and velocity sedimentation studies are interpreted in terms of a single sedimenting species with molecular weights of 21,629 and 21,063 by these respective techniques. These values compare to the predicted molecular weight, from amino acid composition, of 21,817. The hydrodynamic properties of the molecule indicate that it is asymmetric with an axial ratio of 1:5.2 or equivalent dimensions of 21 x 110 A. SCR(1-3) has an unusual CD spectrum exhibiting a broad maximum at 220-230 nm and a minimum at 190 nm. There was little evidence of classical secondary structure. The product exhibited concentration-dependent inhibition of complement-mediated lysis of sensitized sheep red blood cells.


Assuntos
Receptores de Complemento 3b/genética , Receptores de Complemento 3b/isolamento & purificação , Sequência de Aminoácidos , Animais , Dicroísmo Circular , Sequência Consenso , Escherichia coli/genética , Expressão Gênica , Hemólise/efeitos dos fármacos , Humanos , Técnicas In Vitro , Corpos de Inclusão/química , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/isolamento & purificação , Dobramento de Proteína , Receptores de Complemento 3b/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Sequências Repetitivas de Ácido Nucleico , Sefarose , Ovinos , Ultracentrifugação
14.
J Immunol ; 151(3): 1702-11, 1993 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8335953

RESUMO

C receptor type 1 (CR1, CD35) is present in a soluble form in plasma (sCR1). Soluble CR1 was measured with a specific ELISA assay in normal individuals and in patients with different diseases. The mean serum concentration of sCR1 in 31 normal donors was 31.4 +/- 7.8 ng/ml, and was identical in plasma. An increase in sCR1 was observed in 36 patients with end-stage renal failure on dialysis (54.8 +/- 11.7 ng/ml, p < 0.0001), and in 22 patients with liver cirrhosis (158.3 +/- 49.9 ng/ml, p < 0.0001). The mean sCR1 levels dropped from 181 +/- 62.7 to 52.1 +/- 24.0 ng/ml (p < 0.001) in nine patients who underwent liver transplantation, and was 33.5 +/- 7.3 in 10 patients with functioning renal grafts, indicating that the increase in sCR1 was reversible. Soluble CR1 was elevated in some hematologic malignancies (> 47 ng/ml), which included B cell lymphoma (12/19 patients), Hodgkin's lymphoma (4/4), and chronic myeloproliferative syndromes (4/5). By contrast, no increase was observed in acute myeloid or lymphoblastic leukemia (10) or myeloma (5). In two patients with chronic myeloproliferative syndromes, sCR1 decreased rapidly after chemotherapy. The mean concentration of sCR1 was not significantly modified in 181 HIV-infected patients at various stages of the disease (34.8 +/- 14.4 ng/ml), and in 13 patients with active SLE (38.3 +/- 19.6 ng/ml), although in both groups the number of CR1 was diminished on E. There was a weak but significant correlation between sCR1 and CR1 per E in HIV infection and SLE (r = 0.39, p < 0.0001, and r = 0.60, p < 0.03 respectively). In vitro, monocytes, lymphocytes, and neutrophils were found to release sCR1 into culture supernatants. In vivo, sCR1 was detected in the serum of SCID mice populated with human peripheral blood leukocytes. The sCR1 levels correlated with those of human IgG (r = 0.97, p < 0.0001), suggesting synthesis of sCR1 by the transferred lymphocytes. The mechanisms underlining the increased levels of sCR1 and its biologic consequences remain to be defined.


Assuntos
Leucócitos/metabolismo , Receptores de Complemento 3b/metabolismo , Animais , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Eritrócitos/metabolismo , Doenças Hematológicas/sangue , Humanos , Técnicas In Vitro , Falência Renal Crônica/sangue , Cirrose Hepática/sangue , Transplante de Fígado , Camundongos , Camundongos SCID , Receptores de Complemento 3b/química , Solubilidade , Fatores de Tempo , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA